28
Real-time cell cycle imaging in a 3D cell culture model of melanoma Loredana Spoerri 1* , Kimberley A. Beaumont 2,3* , Andrea Anfosso 2,3 , Nikolas K. Haass 1,2,4 1 The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Qld, Australia 2 The Centenary Institute, Newtown, NSW, Australia 3 Sydney Medical School, University of Sydney, NSW, Australia 4 Discipline of Dermatology, University of Sydney, NSW, Australia. * these authors contributed equally to this work Correspondence: Nikolas K. Haass, The University of Queensland Diamantina Institute, Translational Research Institute, 37 Kent St, Woolloongabba, Queensland 4102, Australia Phone +61 7 3443 7087; fax +61 7 3443 6966; e-mail [email protected] Running head: 3D real-time cell cycle imaging

Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

Real-time cell cycle imaging in a 3D cell culture model of melanoma

Loredana Spoerri1*, Kimberley A. Beaumont2,3*, Andrea Anfosso2,3, Nikolas K. Haass1,2,4

1The University of Queensland, The University of Queensland Diamantina Institute,

Translational Research Institute, Brisbane, Qld, Australia

2The Centenary Institute, Newtown, NSW, Australia

3Sydney Medical School, University of Sydney, NSW, Australia

4Discipline of Dermatology, University of Sydney, NSW, Australia.

*these authors contributed equally to this work

Correspondence:

Nikolas K. Haass, The University of Queensland Diamantina Institute, Translational

Research Institute, 37 Kent St, Woolloongabba, Queensland 4102, Australia

Phone +61 7 3443 7087; fax +61 7 3443 6966; e-mail [email protected]

Running head: 3D real-time cell cycle imaging

Page 2: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

i. Summary/Abstract

Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is

an invaluable technique to study cancer cell biology. However, cell cycle progression has

been most commonly assessed by methods that are limited to temporal snapshots or that lack

spatial information. Here we describe a technique that allows spatiotemporal real-time

tracking of cell cycle progression of individual cells in a multicellular context. The power of

this system lies in the use of 3D melanoma spheroids generated from melanoma cells

engineered with the fluorescent ubiquitination-based cell cycle indicator (FUCCI). This

technique allows us to gain further and more detailed insight into several relevant aspects of

solid cancer cell biology, such as tumor growth, proliferation, invasion and drug sensitivity.

ii. Key words: fluorescent ubiquitination-based cell cycle indicator (FUCCI); real-time

imaging; 3D spheroid; tumor heterogeneity; tumor microenvironment; cancer drug resistance;

migration; invasion

1. Introduction

To understand cancer cell biology, especially the process of metastasis, it is necessary to

assess the proliferative and invasive behavior in the context of the location within the tumor

and its complex microenvironment (1,2). In comparison to 2D cell culture models, in vitro

3D tumor models mirror in vivo tumor biology and drug response more faithfully (3-5). They

are therefore an important tool in the field of cancer research and a good compromise

between the lack of a microenvironment encountered under 2D culture conditions and the

great complexity of in vivo animal models.

Page 3: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

Within the range of available in vitro 3D tumor models, spheroids present the advantage of

being the relatively quickest and technically simplest method to recapitulate multiple

characteristics of solid tumors at once: Spheroids mimic physiological tumor behavior in

terms of growth, proliferation, invasion, cell-cell and cell-matrix interactions, molecule

diffusion, oxygen/nutrient gradients – with a hypoxic zone and a central necrosis – as well as

drug sensitivity (3,5). Spheroids can be generated from cell lines but also from primary

patient-derived cells (6).

Since uncontrolled proliferation is a hallmark of malignancies (7,8), understanding cell cycle

behavior in detail is crucial in cancer research. Quantitation of DNA content by flow

cytometry of cells stained with fluorescent markers and DNA pulse incorporation and

detection of nucleotide analogues are probably the most commonly used techniques for cell

cycle analysis. Even though the latter approach provides some chronological insight into the

studied process, both methods provide information limited to snapshots taken at specific time

points. However, cell cycle analysis has been recently revolutionized by a new method:

fluorescent ubiquitination-based cell-cycle indicator (FUCCI) (9). This genetically encoded

system allows spatial and temporal real-time tracking of cell cycle progression of individual

cells in a multicellular context. The FUCCI technology is based on the overexpression of two

modified cell cycle dependent proteins, Geminin and Cdt1, each respectively fused to the

green and red fluorescence emitting proteins Azami Green [mAG-hGem(1-110)] and

monomeric Kusabira Orange 2 [mKO2-hCdt1(30-120)]. Synthesis and degradation of Cdt1

and Geminin during cell cycle progression results in the nucleus of FUCCI-expressing cells

to appear red in G1 phase, yellow in early S phase and green in late S, G2 and M phase.

Immediately following cytokinesis and for a brief period of time at the very beginning of G1

phase the cell nucleus does not display any fluorescence.

Page 4: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

We have incorporated this real-time cell cycle tracking system in to a melanoma 3D in vitro

model by generating spheroids initiated from FUCCI melanoma cell lines (10). We have used

this system to study dynamic heterogeneity in cell cycle behavior and invasion (10), drug

sensitivity (11) and acquired multidrug tolerance in melanoma (12). We have demonstrated

the presence and specific distribution of sub-compartments of cells with different cell cycle

behavior. More specifically, cycling cells were found in the spheroid periphery, while G1-

arrested cells located in more internal zones. This specific distribution correlated with oxygen

and nutrient accessibility. These characteristics reflected those observed in vivo in mouse

xenograft tumors generated from corresponding FUCCI melanoma cell lines, confirming the

physiological relevance of this in vitro cell based model. Furthermore, we showed that

invading cells are actively cycling and that cells arrested in G1, due to their specific location

within the spheroid or to drug treatment, were able to resume proliferation and invasion when

re-exposed to the same favorable conditions experienced by the cycling cells (10).

The method described in this chapter allows us to study the spatiotemporal cell cycle

dynamics of individual cells within the 3D structure of spheroids in real time, and therefore to

gain insight into cancer relevant processes such as proliferation and invasion (13). Briefly,

cultured melanoma cells are transduced with the FUCCI cell cycle indicator system. The red

and green double-positive cells are isolated by fluorescence-activated cell sorting in order to

obtain optimal and comparable fluorescence intensity of both the green and red fluorescence.

Spheroids are then generated using these cells based on a non-adherent surface method

(5,14), embedded in a collagen matrix and their individual cell behavior monitored by

confocal fluorescent time-lapse microscopy. 3D reconstruction of FUCCI cell distribution in

spheroids can be performed using multiphoton microscopy and 3D stitching. The physical

separation of the tumor sub-compartments by Hoechst dye diffusion and subsequent

Page 5: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

fluorescence-activated cell sorting has been described recently (10,14) and is beyond the

scope of this methods chapter.

2. Materials

2.1 Generation of FUCCI expressing melanoma cell lines

1. CO2 incubator.

2. 6-well and 96-well plates.

3. HEK293T cells and the melanoma cell line(s) of choice to be transduced.

4. Distilled H2O (dH2O) and Milli-Q H2O (MQ H2O).

5. Trypsinization solution: 0.05% Trypsin, 0.5 mM EDTA in PBS without Ca2+ and Mg2+.

6. HEK293T medium: 10% fetal bovine serum (FBS) in Dulbecco’s Modified Eagle

Medium.

7. Melanoma cell medium (“Tu4% medium”): 80% MCDB-153 medium, 20% L-15

medium (Leibovitz), 4% FBS, 5 µg/mL insulin, 1.68 mM CaCl2. Dissolve the whole

content of an MCDB-153 vial (17.6 g of powder) in 800 mL deionized H2O, add 15.7 mL

of 7.5% sodium bicarbonate, adjust pH to 7.2-7.4 using NaOH pellets or concentrated

NaOH (see Note 1) and top up with deionized H2O to 1 L. Filter sterilize using a 0.2 µm

filter, remove 200 mL, add 200 mL of L-15 medium (Leibovitz), 40 mL of FBS, 500 µL

of 10 mg/mL insulin and 1120 µL of 1.5 M CaCl2.

8. FUCCI constructs: mKO2-hCdt1 (30-120) and mAG-hGem (1-110) (9), sub-cloned into a

replication-defective, self-inactivating lentiviral expression vector system (15).

9. Lentiviral vectors: pMDLg/pRRE, pRSV-Rev and pCMV-VSV-G (15).

Page 6: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

10. 2× HeBS (HEPES buffered saline): 50 mM HEPES, 10 mM KCl, 12 mM Dextrose, 280

mM NaCl, 1.5 mM Na2HPO4. Adjust to pH 7.1. Filter with a 0.22 µm filter, aliquot and

store at -20°C.

11. 2.5 M CaCl2: To prepare 40 mL, dissolve 11.025 g of CaCl2 in 30 mL of dH2O. Filter

with a 0.22 µm filter, aliquot and store at -20°C.

12. Sterile phosphate buffered solution (PBS): 137 mM NaCl, 2.7 mM KCl, 10 mM

Na2HPO4, 1.8 mM KH2PO4 in H2O.

13. Centrifuge.

14. 0.22 µm and 0.45 µm filters.

15. -80°C and -20°C freezer.

16. Polybrene, 100 mg/mL in sterile H2O. Filter and store at -20°C.

17. Inverted fluorescence microscope.

18. Hemocytometer or automated cell counter.

19. Sorting medium 10% FBS in PBS.

20. FACSAria cell sorter.

21. Inverted phase-contrast microscope.

2.2 Spheroid formation and embedding

1. Well coating solution: 1.5% agarose in sterile PBS. Mix 0.45 g of tissue culture agarose

in 30 mL of sterile PBS (see Note 2) and microwave until the agarose has completely

dissolved (see Note 3). Prepare freshly before plate coating.

2. Trypsin neutralizing medium : 10% FBS in L-15 medium (Leibovitz).

Page 7: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

3. Melanoma cell medium.

4. 200 mM L-Glutamine.

5. 10× Eagle's Minimum Essential Medium (EMEM).

6. Bovine type I collagen (R&D Systems), 5 mg/mL.

7. Fetal bovine serum (FBS).

8. 7.5% (w/v) NaHCO3.

9. Sterile dH2O.

10. 96-well plate.

11. Hemocytometer or automated cell counter.

12. CO2 incubator.

13. Inverted phase-contrast microscope.

2.3 FUCCI-spheroid imaging

1. Time-lapse confocal microscope, e.g. Leica SPF confocal inverted microscope equipped

with 8 laser lines and 5 filter-free PMT detectors (488 and 561 nm lasers to excite mAG

and mKO2), low magnification and long working distance objectives (10× PL FLUOTAR

objective (NA 0.3) or a 20x PL FLUOTAR objective (NA 0.5)) as well as an incubator

chamber to maintain the standard cell culture conditions (i.e. humidity, 37°C and 5%

CO2).

2. Multiphoton microscope, e.g. a custom-built upright TriM Scope two-photon microscope

(LaVision BioTec) equipped with a diode-pumped, wideband mode-locked Ti:Sapphire

femtosecond laser (MaiTai, SpectraPhysics) and an APE Optical Parametric Oscillator

(OPO), Photomultiplier tubes (Hamamatsu Photonics), 520 nm dichroic and bandpass

Page 8: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

filter 560/40 (Chroma Technology) and a water-dipping 20× objective (Olympus

XLUMPlanFL IR coated, NA 0.95).

3. Software: Imaris (Bitplane), Image J/Fiji (open source), Volocity (Perkin Elmer).

3. Methods

3.1 Transfection of HEK293T cells

1. Harvest exponentially growing HEK293T cells (see Note 4) the day before the

transfection, centrifuge at 300 × g for 5 min, count and plate them in a 6-well plate so the

cells will be 70% confluent the next day.

2. On the day of transfection, replace old medium with 2 mL of fresh medium 4 h before

transfection.

3. Prepare 500 µL of calcium phosphate-DNA mix per each transfection in MQ H2O. Co-

transfect HEK293T cells with 0.5 µg each of the packaging defective helper construct

(pMDLg/pRRE), the nuclear localization signal Rev plasmid (pRSV-Rev), a plasmid

coding for a heterologous (pCMV-VSV-G) envelope protein, 8.5 µg of the vector

construct harboring either mKO2-hCdt1 (30-120) or mAG-hGem (1-110) and 50 µL of

2.5 M CaCl2.

4. Add 500 µL of the transfection solution drop by drop to 500 µL of the 2× HeBS solution

while vortexing (see Note  5).

5. Let the solution stand at room temperature for 15 min, vortex every 5 min.

6. Slowly add the 1 mL of solution to each well while gently swirling the plate.

7. After 4 h remove the medium, wash with PBS, add fresh medium and incubate at 37°C,

5% CO2.

Page 9: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

8. After 48 h harvest the supernatant containing the lentiviral particles and centrifuge at 300

× g for 5 min (see Note 6).

9. Filter the supernatant with a 0.45 µm filter.

10. Aliquot in cryovials and store the supernatant at -80°C or proceed with the transduction of

the melanoma cells (see Note 7).

3.2 Transduction of melanoma cells

1. Plate melanoma cell line in a 6-well plate so that they will be in the exponential grow

phase during the transduction the next day (see Notes 8-10).

2. Replace old medium with 2 mL of fresh medium 4 h before transfection.

3. Thaw the lentiviral stock on ice.

4. Prepare the polybrene working solution at 8 mg/mL in culture medium.

5. Prepare 400 µL of transduction solution by mixing 200 µL of each FUCCI lentivirus

(mKO2-hCdt1 (30-120) and mAG-hGem (1-110)) and add 0.4 µl of 8 mg/mL polybrene

(final concentration of 8 µg/mL) (see Notes 11 and 12)

6. Add the transduction solution drop by drop while gently swirling the plate and return

plate to the incubator.

7. After 72 h check the transduction efficiency under the fluorescence microscope.

3.3 Sorting of FUCCI-expressing single cell clones

1. Trypsinize melanoma cells in the exponential growth phase (see Notes 8 and 13).

2. Centrifuge at 300 × g for 5 min.

Page 10: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

3. Remove the supernatant and count the cells.

4. Resuspend the cells in sorting medium at 1 × 106 cells/mL.

5. Using a FACSAria cell sorter, acquire using the 488 nm laser and the B530 detector for

mAG, and B575 for mKO2.

6. Sort double positive cells, highly expressing mKO2-hCdt1 (30-120) and mAG-hGem (1-

100) into a 96-well plate pre-filled with medium.

7. Check at the phase-contrast microscope that no more than one cell is present in each well

(see Note 14).

8. Return to the incubator.

9. Check every day to spot the first dividing clones (see Note 15).

10. Trypsinize cells when confluent and split to a 24-well plate. Continue to expand cells

until there are enough to freeze down in liquid nitrogen for further use.

3.4 Spheroid formation

1. To prepare one 96-well plate for spheroid formation, freshly prepare 30 mL of well

coating solution and immediately dispense 100 µL of the solution in each well (see Note

16). Let the plate rest at room temperature for 15 min to allow the agarose to cool down

and harden.

2. In the meantime, trypsinize the FUCCI-expressing cells (see Note 17): Wash the cells

once with PBS and then treat with 0.05% Trypsin/EDTA for 5 min at 37°C.

3. Neutralize the trypsin by adding trypsin neutralizing medium and pipet repeatedly to

create a single-cell suspension.

Page 11: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

4. Centrifuge at 300 × g for 5 min.

5. Remove supernatant and resuspend the cells in cell culture medium.

6. Count the cells using a hemocytometer or an automatic cell counter and resuspend

600,000 cells in 24 mL of cell culture medium in order to obtain a solution at a

concentration of 2.5 × 104 cells/mL.

7. Using a multichannel pipette add 200 µL of the cell suspension to each well so that 5000

cells are seeded per well (see Note 18).

8. Place the plate in the incubator for the following 72 h, allowing the spheroids to form

(Fig. 1) (see Note 19).

9. Use a phase-contrast microscope to inspect the spheroids and mark the wells where the

spheroids have formed properly (see Note 20).

[Fig. 1 near here]

3.5 Spheroid embedding in collagen matrix

1. To prepare 1 mL of the collagen embedding solution, mix 100 µL of 10X EMEM, 10 µL

of 200 mM L-Glutamine and 400 µL of bovine type I collagen (see Note 21 and Note

22). Work on ice (see Note 23).

2. Titrate the solution with 7.5% NaHCO3 until the solution turns from a yellow-orange to a

peach-pink color (the volume required for this change in pH should be around 15-30 µL).

3. Add 100 µL of FBS and top up to 1 mL with H2O.

4. Mix well while taking care to avoid generating bubbles and keep on ice.

Page 12: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

5. Add 40 µL in each well of a new flat bottom 96-well plate (see Note 24) and incubate

plate for 5 min at 37°C to allow the collagen to polymerize (See Note 25).

6. Transfer spheroids from the 96-well plate where they have been cultured to sterile

microcentrifuge tubes using a pipette and pipette tips with enlarged aperture (see Note

26). Transfer one spheroid per tube (Fig. 2) (see Note 27).

7. Let spheroids settle by gravity to the tube’s bottom and remove the supernatant taking

care to not disturb the spheroid (see Note 28).

8. Gently resuspend each spheroid in 60 µL of collagen solution (use pipette and pipette tips

with enlarged aperture) and transfer into the 96-well plate that was previously coated with

40 µL per well of collagen solution (step 5).

9. Incubate plate for 15 min at 37°C to allow the collagen to polymerize.

10. With the use of a phase-contrast microscope inspect spheroid integrity and distribution in

the well (if multiple spheroids were seeded per well).

11. Add 200 µL per well of appropriate medium and incubate at 37°C until the setup of the

spheroid time-lapse confocal microscopy analysis.

[Fig. 2 near here]

3.6 FUCCI-spheroid time-lapse imaging using confocal microscopy

1. Set the microscope to a low magnification and turn on the laser to excite the Azami Green

or Kusabira Orange 2 proteins (see Note 29).

2. Move to the well containing the spheroid to be imaged, locate the spheroid and center it

in the field of view.

Page 13: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

3. Set the microscope to a magnification that allows visualization of the whole spheroid (see

Note 30).

4. Position the plane of view in the center of the spheroid (see Note 31). To do so, adjust the

z variable starting with the view plane at the bottom of the spheroid then moving it deeper

into the spheroid. During this process the diameter of the spheroid should increase until

the center of the spheroid has been reached (see Note 32).

5. Adjust the necessary settings (exposure time, gain, laser power, etc.) to achieve optimal

signal without overexposed zones.

6. Set up four optical slices on each side of the central plane, each separated by 5 µm. This

results in a total of 9 optical slices (including the central slice) encompassing a thickness

of 40 µm, 20 µm above the central plane and 20 µm below the central plane of the

spheroid (see Note 33).

7. Set up the total recording duration to at least 24-48 h and the intervals’ time to 10-20 min

(see Note 34).

8. Start the time-lapse recording (Fig. 3).

9. The obtained images are processed using Imaris, Volocity or Image J. Obtain cell

parameters such as speed by automated or manual cell tracking in the aforementioned

software programs. Convert time-lapse image sequences into AVI or MOV format for

display in common media players.

[Fig. 3 near here]

3.7 Spheroid imaging using multiphoton microscopy and 3D stitching

Page 14: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

1. Visualize the spheroid using a 20x water-dipping objective, lowered into the medium (see

Note 35).

2. Excite the FUCCI-spheroid using a wavelength of 920 nm (Ti:Sapphire laser) and 1060

nm (OPO).

3. Use a 465 dichroic to remove second-harmonic generation (SHG) signals originating

from the Ti:Sapphire laser (see Note 36).

4. Use a 520 nm dichroic to split the mKO from the mAG emission, and a bandpass filter

560/40 to further refine the mKO signal and exclude SHG light originating from the

OPO.

5. Collect mAG and mKO fluorescence in photomultiplier tubes.

6. Acquire three-dimensional images (x, y, z) of spheroids using the LaVision acquisition

software. Use a z spacing of 4 µm, with montaging to cover the entire XY area of the

spheroid. Use a z-depth of approximately 600 µm or until the signal fades, in order to

image as much of the spheroid depth as possible.

7. Stitch z-stack images in FIJI or ImageJ using the Stich Grid of Images Plugin (16), and

perform 3D volume rendering using Volocity software (Fig. 4) (see Note 37).

[Fig. 4 near here]

4. Notes

1. To start pH should be around 6.8 and should need 2-3 NaOH pellets to reach the right pH

and change color to red. Notice that pH might increase slightly when topping up with

H2O to 1 L.

Page 15: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

2. It is advisable to prepare at least 20 mL extra in addition to the required volume. Due to

its viscosity this solution tends indeed to stick to the surfaces of the container.

3. Coating plates with 1.5% agarose will prevent adhesion of the cells to the bottom of the

well and at the same time creates a meniscus that allows grouping of the cells at the

center of the well.

4. HEK293T is the embryonic kidney 293 cell line (HEK293) carrying the SV40 T-antigen.

The T-antigen makes these cells highly transfectable with vectors carrying the SV40

origin of replication resulting in high viral titers.

5. Add the 2× HeBS drop-wise to the CaCl2/plasmid DNA solution, to achieve optimal

calcium-phosphate/DNA precipitate.

6. Gloves and lab coat must be used when working with lentiviral vectors. The transduction

must be carried out in a PC2 laboratory equipped with a certified Class II Biosafety

cabinet and a tissue culture incubator. The buckets in the centrifuge must be equipped

with aerosol-tight covers. Decontaminate the surface of the Biosafety cabinet with 10%

bleach at the end of the session.  

7. Lentiviral particles can be used fresh, or stored at -80˚C. After thawing and re-freezing,

there will be a loss of virus potency.

8. The number of cells to be seeded depends on the cell line used.

9. This step is very important as actively dividing cells give a higher transduction rate than

non-dividing cells.

10. Low cell confluence is desired to allow growth for 2 days without splitting.

Page 16: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

11. Remember to perform also the single color and the mock transduction as later flow

cytometry sorting will be performed for which single color signal compensation is

required.

12. Calculation of the virus MOI is not required as expression levels of each construct can be

measured by flow cytometry and used for sorting to achieve comparable fluorescence

intensity of the FUCCI fluorophores.

13. This is important because confluent cells arrest in G1 and accumulate Cdt1. This will

result in stronger intensity of the red fluorescence compared to actively dividing cells that

pass through G1.

14. It is possible that no cell is visible at this stage.

15. Usually, cells grow close to the edge of the wells. Two cells close together are likely a

single cell clone. Two cells far apart within the well are likely two independent clones

and should therefore be disregarded.

16. This procedure needs to be performed quickly in order to prevent hardening of the

agarose before it has been dispensed in the wells. Use a multichannel pipette and change

tips between each 96-well plate (if preparing multiple plates).

17. In order to facilitate easier cell tracking, untransduced parental cells (non-fluorescent)

may be mixed with FUCCI cells in a ratio of 10:1 or even 20:1 when forming spheroids.

This will reduce problems with cell separation in imaging software due to density.

18. It is advisable to use cells that were grown at a density of 80% or lower. We have

observed in the past that cells at higher confluency resulted in poor spheroid formation.

19. The cells will initially group in close contact to each other as a non-adherent single layer

in the center of the well. Over time they will initiate cell-cell contact and begin to form a

Page 17: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

3D structure. Once cell-cell contact is initiated (usually after 24 h), the cells will re-start

to proliferate allowing the spheroid to expand.

20. A properly formed spheroid should have a reasonably round and regular shape with a

necrotic core observable by phase-contrast microscopy as a darker central zone, although

the latter might not be visible in all cell lines. The success of spheroid formation depends

on the specific cell lines used to form spheroids as well as on the integrity of the agarose

coating, i.e. spheroids can form properly in some well but not in others. Some cell lines

require seeding of higher number of wells compared to others in order to obtain the same

number of properly formed spheroids.

21. The volumes given here are for 1 mL of collagen solution which is enough to embed 9

spheroids, each in a well of a 96-well plate (100 µL per spheroid plus minimal

manipulation-related loss of volume). To embed spheroids in larger wells (48- or 24-well

plates), adjust the volumes proportionally to the well’s surface area. The number of

spheroids embedded per well can also be adjusted accordingly (e.g. 3 spheroids in a well

of a 24-well plate), however take into consideration that multiple spheroids seeded in a

single well might embed in close contact to each other, preventing/compromising their

experimental use.

22. In addition, other cell types such as fibroblasts and/or endothelial cells can be added to

the collagen gel to mimic the stroma better. Examples of this technique are outlined in

(17-19).

23. All work must be undertaken on ice in order to avoid collagen polymerization, unless

otherwise desired, i.e. during and following incubation at 37°C.

Page 18: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

24. A normal plastic tissue culture plate may be used - but in order to improve image quality,

and if a smaller working distance objective is needed, an imaging plate (either glass or

thin plastic bottom) should be used.

25. This is important to prevent contact between the transferred spheroid and the bottom of

the well. Contact will allow cells that are part of the spheroids to adhere to the plastic and

proliferate as a monolayer on the well’s surface.

26. With a pair of scissors, enlarge the opening of the pipette tip by cutting approximately 2-3

mm off the tip. Before use, clean and disinfect the blades of the scissors with 80%

ethanol.

27. To successfully aspirate a spheroid, immerse the end of the pipette tip in the supernatant

and move as close as possible to the spheroid without touching it, then gently but

assertively retract the plunger to generate a fluid flow with which the spheroid will move

into the tip. Gently release the spheroid into the tube avoiding to damage it.

28. Remove the majority of the supernatant using a pipette attached to a vacuum pump then

carefully remove as much as possible of the remaining supernatant using a manual pipette

with a gel-loading tip.

29. In the case of untreated spheroids the green and red fluorescence signals are expected to

have similar intensity and their use to locate the spheroid is interchangeable. If the

intensity of one of the two fluorescence signals is decreased (e. g. due to drug treatment),

the channel for the fluorophore with the strongest intensity should be used to locate the

spheroid.

30. The starting size and growth rate of the spheroid, as well as whether visualization of cell

invasion is desired, are factors to consider when choosing the magnification. If

visualization of cell invasion of a fast growing spheroid is desired, the visualization field

Page 19: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

at the start of the experiment should include sufficient empty space surrounding the

spheroid accounting for the increase in size of the spheroid as well as cell moving out of

the spheroid.

31. The ideal position to monitor cell cycle behavior in FUCCI melanoma spheroids is in the

middle of the spheroid, as this allows to observe both the inner arrested zone and the

outer cycling layer of cells (based on the specific distribution of sub-compartments of

cells with different cell cycle behavior outlined in the introduction paragraph).

32. Due to the limited sample penetration depth of confocal microscopy, it might not be

possible to visualize the central plane of certain spheroids without experiencing loss of

signal. This depends on the spheroid’s size, density as well as other factors. The loss of

signal might be uneven throughout the plane and give rise to artifacts. The plane of view

should be localized as deep as possible without experiencing loss of signal. This

compromise might be adopted where setting the plane of view slightly away from the

center of the spheroid still provides the desired information. To confirm the lack of

artifacts, the same experiment can be repeated for specific time points where sections of

the fixed spheroid (instead of the whole spheroid) are imaged using the same microscopy

technique. This method prevents potential artifacts generated by limited imaging

penetration depth.

33. Collecting z-stack images allows the creation of a maximum intensity projection image to

enhance signal intensity when signal from a single optical slice is too weak. The thickness

of the z-stack and the number of optical slices it contains should be adjusted according to

spheroid’s characteristics and in particular to spheroid size. For instance, in melanoma

FUCCI spheroids, an excessive z-stack thickness might result in signal contamination

from the outer cycling layer into the inner arrested zone of cells. Removal of the bottom

z-slices is therefore necessary for optimal outcome.

Page 20: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

34. These factors depend on the time necessary for the phenomenon of interest to be captured

and the desired level of details. For monitoring changes in cell cycle behavior using the

FUCCI system the total recording duration and intervals’ time need to be adjusted

according to the cell line’s cell cycle time.

35. An inverted multiphoton microscope may be preferable if repeated imaging is to take

place, or time lapse imaging. This will avoid potential contamination due to the need to

remove the tissue culture plate lid.

36. SHG may also be used to image type I collagen fibers (20,21).

37. Several other imaging software have 3D stitching plugins, and FIJI also has additional

stitching plugins. If this FIJI plugin fails, one of the other methods should be tried. An

alternative is to start with two tiles that do stitch together, and then to build up the larger

image iteratively by gradually adding more tiles. For this method the maximum intensity

blending method must be used.

Acknowledgements

We thank Prof Meenhard Herlyn, The Wistar Institute, Philadelphia, PA; Prof Keiran

Smalley, Moffitt Cancer Center, Tampa, FL; Prof Wolfgang Weninger & Dr Ben Roediger,

Centenary Institute, Sydney, NSW; Dr Crystal Tonnessen, UQ Diamantina Institute,

Brisbane, QLD, and the imaging facilities of the Centenary and UQ Diamantina Institutes for

their contribution to optimizing this protocol over the years. We thank Prof Atsushi

Miyawaki, RIKEN, Wako-city, Japan, for providing the FUCCI constructs. N.K.H. is a

Cameron fellow of the Melanoma and Skin Cancer Research Institute, Australia. K.A.B. is a

fellow of Cancer Institute New South Wales (13/ECF/1-39). The work leading to this

protocol was supported by project grants RG 09-08 and RG 13-06 (Cancer Council New

Page 21: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

South Wales), 570778 and 1051996 (Priority-driven collaborative cancer research

scheme/Cancer Australia/Cure Cancer Australia Foundation), 08/RFG/1-27 (Cancer Institute

New South Wales), APP1003637 and APP1084893 (National Health and Medical Research

Council).

5. References

1. Brandner JM, Haass NK (2013) Melanoma's connections to the tumour microenvironment.

Pathology 45 (5):443-452. doi:10.1097/PAT.0b013e328363b3bd

2. Villanueva J, Herlyn M (2008) Melanoma and the tumor microenvironment. Curr Oncol

Rep 10 (5):439-446

3. Beaumont KA, Mohana-Kumaran N, Haass NK (2014) Modeling Melanoma In Vitro and

In Vivo. Healthcare 2 (1):27-46. doi:10.3390/healthcare2010027

4. Santiago-Walker A, Li L, Haass NK et al. (2009) Melanocytes: from morphology to

application. Skin Pharmacol Physiol 22 (2):114-121

5. Smalley KS, Lioni M, Noma K et al. (2008) In vitro three-dimensional tumor

microenvironment models for anticancer drug discovery. Expert Opin Drug Discov 3 (1):1-

10. doi:10.1517/17460441.3.1.1

6. Wroblewski D, Mijatov B, Mohana-Kumaran N et al. (2013) The BH3-mimetic ABT-737

sensitizes human melanoma cells to apoptosis induced by selective BRAF inhibitors but does

not reverse acquired resistance. Carcinogenesis 34 (2):237-247. doi:10.1093/carcin/bgs330

7. Hanahan D, Weinberg RA (2000) The hallmarks of cancer. Cell 100 (1):57-70

8. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144

(5):646-674

Page 22: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

9. Sakaue-Sawano A, Kurokawa H, Morimura T et al. (2008) Visualizing spatiotemporal

dynamics of multicellular cell-cycle progression. Cell 132 (3):487-498

10. Haass NK, Beaumont KA, Hill DS et al. (2014) Real-time cell cycle imaging during

melanoma growth, invasion, and drug response. Pigment Cell Melanoma Res 27 (5):764-776.

doi:10.1111/pcmr.12274

11. Beaumont KA, Hill DS, Daignault SM et al. (2016) Cell cycle phase-specific drug

resistance as an escape mechanism of melanoma cells. J Invest Dermatol.

doi:10.1016/j.jid.2016.02.805

12. Ravindran Menon D, Das S, Krepler C et al. (2015) A stress-induced early innate

response causes multidrug tolerance in melanoma. Oncogene 34 (34):4448-4459.

doi:10.1038/onc.2014.372

13. Haass NK (2015) Dynamic tumor heterogeneity in melanoma therapy: how do we address

this in a novel model system? Melanoma Manag 2 (2):93-95

14. Beaumont KA, Anfosso A, Ahmed F et al. (2015) Imaging- and Flow Cytometry-based

Analysis of Cell Position and the Cell Cycle in 3D Melanoma Spheroids. J Vis Exp

106:e53486. doi:10.3791/53486

15. Smalley KS, Brafford P, Haass NK et al. (2005) Up-regulated expression of zonula

occludens protein-1 in human melanoma associates with N-cadherin and contributes to

invasion and adhesion. Am J Pathol 166 (5):1541-1554

16. Preibisch S, Saalfeld S, Tomancak P (2009) Globally optimal stitching of tiled 3D

microscopic image acquisitions. Bioinformatics 25 (11):1463-1465.

doi:10.1093/bioinformatics/btp184

17. Flach EH, Rebecca VW, Herlyn M et al. (2011) Fibroblasts contribute to melanoma

tumor growth and drug resistance. Mol Pharm 8 (6):2039-2049. doi:10.1021/mp200421k

Page 23: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

18. Haass NK, Sproesser K, Nguyen TK et al. (2008) The mitogen-activated

protein/extracellular signal-regulated kinase kinase inhibitor AZD6244 (ARRY-142886)

induces growth arrest in melanoma cells and tumor regression when combined with

docetaxel. Clin Cancer Res 14 (1):230-239

19. Velazquez OC, Snyder R, Liu ZJ et al. (2002) Fibroblast-dependent differentiation of

human microvascular endothelial cells into capillary-like 3-dimensional networks. Faseb J 16

(10):1316-1318

20. Kirkpatrick ND, Hoying JB, Botting SK et al. (2006) In vitro model for endogenous

optical signatures of collagen. J Biomed Opt 11 (5):054021. doi:10.1117/1.2360516

21. Tong PL, Qin J, Cooper CL et al. (2013) A quantitative approach to histopathological

dissection of elastin-related disorders using multiphoton microscopy. Br J Dermatol 169

(4):869-879. doi:10.1111/bjd.12430

Page 24: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique

   

Figure legends

Figure 1. Spheroid formation: A day-by day schematic illustration of the different stages of

spheroid formation and the main steps involved in this procedure.

 

Figure 2. Spheroid embedding: schematic illustrating the main steps involved in the

procedure for embedding spheroids in collagen matrix.

 

Figure 3. Confocal extended focus image of a FUCCI-melanoma spheroid (1:10 FUCCI-

C8161 cells to wt-C8161 cells, from 0-13 h after collagen embedding. The top few z-slices

were removed to reveal the red G1-arrested cells inside the spheroid. White arrows indicate a

single invading cell tracked over time. Note the cell cycle change of the invading cell from

G1 (red) to S/G2/M (green). Scale bar, 100 µm.

 

Figure 4. 3D rendering of a multiphoton microscopy image z-stack of a FUCCI-melanoma

spheroid (C8161) 24 h after collagen implantation.

Page 25: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique
Page 26: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique
Page 27: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique
Page 28: Real-time cell cycle imaging in a 3D cell culture model of ... · Aberrant cell cycle progression is a hallmark of solid tumors; therefore cell cycle analysis is an invaluable technique